Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2014 Mar 30;71(16):3101–3117. doi: 10.1007/s00018-014-1607-2

Tracking migration during human T cell development

Joanna Halkias 1,2,, Heather J Melichar 1, Kayleigh T Taylor 1, Ellen A Robey 1
PMCID: PMC11113765  PMID: 24682469

Abstract

Specialized microenvironments within the thymus are comprised of unique cell types with distinct roles in directing the development of a diverse, functional, and self-tolerant T cell repertoire. As they differentiate, thymocytes transit through a number of developmental intermediates that are associated with unique localization and migration patterns. For example, during one particular developmental transition, immature thymocytes more than double in speed as they become mature T cells that are among the fastest cells in the body. This transition is associated with dramatic changes in the expression of chemokine receptors and their antagonists, cell adhesion molecules, and cytoskeletal components to direct the maturing thymocyte population from the cortex to medulla. Here we discuss the dynamic changes in behavior that occur throughout thymocyte development, and provide an overview of the cell-intrinsic and extrinsic mechanisms that regulate human thymocyte migration.

Keywords: Human thymocytes, Migration, Chemokines, Integrins, Extracellular matrix, T cell receptor

Introduction

The development of T cells capable of providing immunity against a diverse set of microbial and environmental antigens must be carefully balanced with deletion of potentially auto-reactive cells. The generation of these functionally mature, self-tolerant T cells occurs within anatomically and functionally distinct regions of the thymus, each containing specialized niches that regulate the sequential stages of thymocyte differentiation and maturation (Fig. 1). Thus, the complex and carefully coordinated journey of thymocytes through the thymus is essential for proper T cell development. Mechanistic insight into the events that direct thymocyte migration could guide the development of strategies to manipulate the T cell repertoire in the treatment of immune-mediated diseases or to increase thymic output and improve immune reconstitution following bone marrow (BM) transplantation.

Fig. 1.

Fig. 1

Traffic of thymocytes in the human thymus. Boxed area represents an enlarged cortico-medullary region of the human multi-lobular thymic structure. a Circulating thymus-colonizing hematopoietic progenitor cells arrive via the vasculature and enter the postnatal thymus at the cortico-medullary junction. b Early progenitors commit to the T cell lineage and differentiate to DN thymocytes as they migrate through the cortex. c Those DN cells which productively rearrange the TCR β gene then proceed to up-regulate CD4 and CD8 and progress to a DP stage. DP thymocytes scan the cortex and interact with cTEC and cortical dendritic cells and proceed through positive and negative selection. d Positively selected DP thymocytes that have received a low-affinity TCR signal differentiate into SP thymocytes and migrate to the medulla. e The process of negative selection continues in the medulla, where thymocytes with high avidity for tissue-restricted antigens are deleted. f Mature SP thymocytes exit the thymus and enter the circulation

Structurally, the thymus is divided into an outer cortex and an inner medulla, two functionally distinct regions with specific contributions to the differentiation of T cells. BM-derived thymus-seeding progenitor cells arrive predominantly via the vasculature and enter the cortex, where initial T cell commitment at the CD4CD8 (double negative, DN) progenitor stage occurs (Fig. 1a, b). Following the productive rearrangement of the TCR β chain and the developmental transition to the CD4+CD8+ (double positive, DP) stage, the process of positive selection ensures the development of T cells with functional αβ T cell receptors (TCRs) (Fig. 1c, d). Cortical thymic epithelial cells (cTECs) have distinct antigen-processing capabilities that result in the generation of a unique peptide repertoire optimized for positive selection [13]. During positive selection, DP thymocytes scan the unique repertoire of self-peptides presented by cTECs in the context of MHC, and those DP cells with TCRs that engage in low-affinity interactions with these self-peptide/MHC complexes go on to become CD4+ or CD8+ single positive (SP) thymocytes. The transition from the DP to SP stage is accompanied by the relocalization of thymocytes from the cortex to the thymic medulla (Fig. 1e). While there is evidence that negative selection of thymocytes bearing TCRs with high-avidity for ubiquitous self-antigens begins in the cortex [4], the thymic medulla is necessary for the deletion of additional self-reactive thymocytes. The specialized medullary microenvironment contains a higher concentration of dendritic cells (DCs) to present self-antigens for negative selection, and medullary thymic epithelial cells (mTECs) that express tissue-restricted antigens under the control of the transcription factor autoimmune regulator (AIRE) [5]. Thymocytes that successfully complete these stringent TCR selection events are exported from the thymus as mature CD4+ and CD8+ SP T cells to perform unique effector functions in host immune defense (Fig. 1f). Defective thymocyte migration at any of these stages of T cell development can result in lymphopenia or autoimmune disease.

Much of what we know about thymocyte development and migration is based on studies in mice, but to what degree this is applicable to humans is not known. While there are many similarities between mouse and man, there are also salient differences, highlighting the need for the direct study of human T cell development. In this review, we will provide an overview of human thymocyte migration and how it impacts the organization of the human thymus, the differentiation of human T cells, and its role in human disease.

Models to study human thymocyte migration

Exploration of the molecular mechanisms underlying human primary immune deficiencies has led to the identification of many of the signaling cues involved in human T cell development and migration, and modeling these deficiencies in mice remains a powerful tool for the study of immune development. Thymic tissue is rarely available from immunodeficient patients. However, normal human thymic issue can be obtained from fetal products of conception or as a surgical byproduct during pediatric cardiac surgery. These tissues have provided invaluable information regarding the phenotype of human thymocytes and their compartmentalization during the different stages of development. Immunostaining of fixed human thymic tissue has enabled the characterization of human epithelial cells and DCs within the cortex and medulla and has also provided some information on the patterns of chemokine expression that direct human thymocyte migration within the thymus. However, technical challenges associated with immunostaining for secreted proteins such as chemokines have yielded conflicting results. For example, fluorescent and immunohistochemical staining suggests CXCL12 is expressed along the cortico-medullary junction (CMJ) or in the medulla of the human thymus [6, 7]. In contrast, gene expression profiles of human and murine purified thymic stromal cell populations clearly indicate higher CXCL12 expression in cortical versus medullary thymic epithelial cells [8, 9].

A number of in vitro assays have been developed to study human cell migration, each representing a simplified approximation of in vivo conditions [10]. In vitro transwell and two-dimensional migration studies have identified candidate chemokines and adhesion molecules that could potentially play a role in guiding human thymocyte migration at distinct stages of development (summarized in Tables 1, 2). Although it is well established that chemokine concentration gradients can direct cell migration in vitro, only recently has it been shown that the migration of DCs in vivo is dependent on a functional chemokine gradient [11]. While in vivo evidence is lacking for human T cells, the development of 3D chemotaxis assays for live imaging of cell movement within a collagen matrix has allowed for the direct visualization of human T cell migration along a concentration gradient [12]. The migration of human thymocytes in vivo is likely governed by the integration of a multitude of different, and possibly opposing, signaling cues, whose additive effect determines the overall migration pattern of thymocytes at a given stage of development. Thus, these data should be viewed in context with data from complementary systems to study human thymocyte migration in situ.

Table 1.

Differential expression of integrins implicated in thymocyte migration

Integrins Expression of integrins by thymocyte subset References
DN Pre-selection DP Post-selection DP SP
α4β1 +++/adhesion ++/adhesion ++/migration +/migration [106, 107, 118, 119]
α5β1 +++/adhesion ± ++/migration ++/migration [106, 107, 118, 119]

Table 2.

Differential expression of chemokines and chemokine receptors implicated in human thymocyte migration

Chemokine receptors Expression of chemokine receptors by thymocyte subset References Ligands Location of ligand expression References
DN Pre-selection DP Post-selection DP SP
CXCR4 +++F, P +++F, P +F, P ++F, P [16, 102, 103, 112, 113] CXCL12/SDF-1 Cortex M [9]
CCR7 +F, P −F, P +F, P ++/+++F, P [16, 62] CCL19/MIP-3 beta, ELC, exodus-3, CK beta 11 Medulla S [62]
CCL21/6Ckine, Exodus-2, SLC, TCA-4, CK beta 9 Medulla S [20]
CCR9 +F +++F +++F ++F [16, 104, 105] CCL25/TECK, CK beta 15 Cortex S [20]
CCR4 +F, Ha +F, Ha ++F, H [62] CCL22/MDC Medulla S [62, 63]
CCR3 −F, P +F, Pa +F, Pa ++P, F [65] CCL11/Eotaxin Medulla S [65]
CXCR3 +F, P −F, Pa −F, Pa ++F, P [64] CXCL9/MIG Medulla H, S [64]
CXCL10/IP-10 Subcapsular cortex S, medulla H, S [64]
CXCL11/I-TAC Medulla H, S [64]

The method of detection is indicated by: F functional chemotaxis assays, P protein expression by flow cytometry, S immunohistochemical or fluorescent antibody staining of sections, H in situ hybridization, M gene expression profiles by microarray analysis

CCR chemokine receptor, CXCR chemokine receptor, CCL chemokine ligand, CXCL chemokine ligand

aResults from DP population as a whole, no distinction between pre- and post-selection DP thymocytes

To this end, several models to study human thymocyte migration in situ have been developed. Studies using chimeric fetal thymic organ cultures (FTOC), in which human progenitors are seeded onto murine fetal thymic lobes that can be maintained in culture for several weeks, have yielded fundamental information regarding the identity of human thymus-seeding progenitors and their differentiation potential, and have also helped characterize the stages of human thymocyte differentiation [13]. These interspecies organotypic culture systems allow for use of an expanded toolkit of the many genetic manipulations available in mice, but have the disadvantage that not all receptor:ligand interactions between species are conserved. Alternately, FTOCs composed of minced human thymic fragments provide a three-dimensional human thymic environment [14, 15]. However, these fragments do not necessarily recapitulate the subcapsular regions of the cortex nor do they reflect the cortico-medullary compartmentalization of a mature thymus. More recently, we have adapted a thymic slice model in which the cortical/medullary architecture is maintained, and in conjunction with two-photon microscopy, enables us to examine the dynamics of human thymocyte motility and behavior in their native tissue [16]. While human thymic organotypic culture systems provide excellent models in which to study human thymocyte migration, the lack of vascular or lymphatic connections makes them less suitable for examining thymic entry or egress.

Humanized mice, in which human hematopoietic progenitors give rise to a human immune system, could provide an important system to study certain aspects of human thymocyte migration that are best addressed in vivo. However, significant sequence divergence between mouse and human cytokines greatly contributes to the decreased cellularity and limited reconstitution of the murine thymus in humanized mice, and there are likely other inefficiencies in interspecies receptor:ligand interactions that further complicate this system [1719]. Therefore, although humanized mice may represent an improvement over existing models, important caveats remain when considering the suitability of humanized mice as a model for the study of some aspects of human thymocyte development and migration.

In sum, varied and complementary systems ranging from single-cell suspension assays to organotypic cultures and humanized mice have been creatively designed to dissect the signaling cues guiding human thymocyte development and migration. Each of these models presents their own set of advantages and limitations, but if carefully applied, can provide a diverse set of tools to dissect the signaling cues directing the journey of developing human T cells through the thymus. As we review the literature, we will point out limitations that remain and challenges that lie ahead as we investigate the different aspects of human thymocyte migration.

Organization of the thymus

Thymic organogenesis

Several aspects of basic thymus anatomy differ in mice and humans. For example, the multi-lobular organization of the human thymus is in contrast to the bi-lobed thymus of mice, and there are well-developed Hassall’s corpuscles in the human but not the murine thymus. Despite these differences, both the murine and human thymus develop exclusively from the third pair of pharyngeal pouches, and many of the genes involved in thymus organogenesis are conserved between the two species [20]. Following detachment from their site of origin in the pharynx, the epithelial primordia of mammals migrate ventrally to fuse at the pericardium. During murine embryogenesis, the migration of the thymus to the anterior chest occurs at approximately embryonic day 11.5 (e11.5), concurrent with the appearance of the first hematopoietic progenitors and prior to the formation of blood vessels [21]. Similarly, in humans, the thymic anlage reaches its final position in the body by the 8th week of gestation, along with the concomitant colonization of the tissue by lymphoid progenitors and prior to the establishment of the thymic vasculature [20, 22, 23]. Human TEC differentiation follows the onset of Foxn1 expression around week 8, resulting in the segregation of cTECs and mTECs and the establishment of a distinct cortico-medullary boundary by weeks 13–16 [2024]. Further maturation of the medulla is evidenced by the onset of AIRE expression in mTECs by week 13–14, and the appearance of Hassall’s corpuscles by 14–15 weeks of gestation [22]. Defects in Foxn1 expression lead to thymic aplasia and the development of severe combined immune deficiency (SCID) in both humans and mice [2527], while an absence of AIRE expression results in severe autoimmunity [5, 28, 29], underscoring the conservation of gene expression and function in thymic organogenesis between the two species.

Notwithstanding remarkable similarities in thymus organogenesis, the ontogeny of T cell development differs significantly between species. The appearance of TCRγδ+ thymocytes precedes that of TCRαβ+ cells in the murine fetal thymus, whereas αβ and γδ T cells emerge simultaneously during human fetal thymopoiesis [23, 30, 31]. Additionally, murine T cells are not evident in the periphery until e15, while the appearance of mature thymocytes occurs comparatively earlier in human development, with seeding of peripheral organs by mature naïve T cells as early as 12–14 weeks of gestation [22, 32, 33]. Perhaps one of the most striking differences between murine and human fetal T cell development is the appearance of human Treg cells in the periphery concurrently with the appearance of naïve T cells, while murine Treg cells are not detected in the periphery before day 3 of life [3437].

There is ample evidence in mice that the normal differentiation of cortical and medullary epithelial cells, as well as formation of a mature medullary compartment during organogenesis, depends on thymocyte-derived signals [3843]. Strong evidence for thymocyte:epithelial cell cross talk is also evident from the thymic biopsies of patients with severe combined immune deficiencies (SCID) caused by mutations in the genes encoding for the IL2 receptor gamma chain (IL2Rγ), JAK3, and adenosine deaminase, among others. These defects in the early stages of human T cell development result in profound abnormalities of TEC differentiation, loss of cortico-medullary demarcation, and an absence of AIRE+ mTECs and Hassall’s corpuscles [44]. For example, Omenn syndrome presents with the unique combination of immunodeficiency and autoimmunity. Although the primary defect is a hypomorphic mutation in recombination-activating genes resulting in impaired VDJ recombination and an oligoclonal T cell repertoire [45, 46], the expansion of these oligoclonal self-reactive T cells in the periphery has been suggested to result from defective negative selection [47]. Indeed, the thymi of patients with Omenn syndrome have markedly reduced AIRE+ mTECS, and these findings are replicated in a mouse model of Omenn syndrome, suggesting that impaired negative selection due to absent AIRE expression may allow the escape of self-reactive T cells [4749]. Treatment of SCID or Omenn syndrome patients with BM transplantation results in thymus-dependent T cell reconstitution and development of a polyclonal T cell repertoire [50, 51]. Similarly, reconstitution of the thymic rudiment in Rag−/−IL-2Rγ−/− mice with human CD34+ progenitor cells results in the formation of a well-defined medulla containing differentiated mTECs [18].

The thymic microenvironment

Thymic epithelial cells are the most abundant stromal cells of the thymus and define anatomically and functionally distinct regions involved in directing the step-wise differentiation of thymocytes. Specialized epithelial cells of the thymic cortex provide positive selection signals, while the medulla contains a unique epithelial cell population with “promiscuous” expression of tissue-restricted antigens. The human thymic medulla also contains well-developed Hassall’s corpuscles composed of keratinized epithelial swirls, which have been implicated in the development of human regulatory T (Treg) cells [52]. In addition to epithelial cells, the human and murine thymus also contains BM-derived hematopoietic cells. These include antigen presenting cells such as DCs, involved in negative selection events [4, 53, 54], and macrophages, implicated in the clearance of dying thymocytes [5557].

In addition to their role in thymocyte selection, thymic epithelial cells help produce another important part of the thymic microenvironment––the extracellular matrix (ECM). Generally, the ECM is composed of combinations of multiple collagens, and several isoforms of both laminin and fibronectin [58]. The ECM network is well preserved between humans and mice, and while extensive in the medulla, it is composed of only fine fibers in the cortex [59]. ECM components are important for the integrity of the tissue, the division of the thymus into lobules, and additionally provide developing T cells cues to proliferate, differentiate, and migrate. Accordingly, thymocytes demonstrate developmentally regulated expression of ECM specific receptors such as integrins, trans-membrane receptors that mediate cell:cell and cell:ECM adhesion (Table 1). Thymocyte motility depends on the regulation of repeated cycles of integrin-mediated adhesion and de-adhesion, along with cytoskeletal rearrangements and the generation of cell polarity. Essential to the high degree of flexibility required for cell adhesion during motility is the ability to alter the avidity of integrins for their ligands through an intra-cellular signaling process known as inside-out signaling [60].

The interactions of thymocytes with their microenvironment are also controlled by chemokines, a family of secreted proteins that induce chemotaxis. Traditionally, chemokines and their receptors are divided into four subfamilies (CC, CXC, C, or CX3C), identified according to the pattern of cysteine residues in the ligands. Each chemokine can bind one or more trans-membrane G-protein coupled chemokine receptors, and in turn, each chemokine receptor can bind many ligands, resulting in a functional redundancy that attenuates the phenotype of many mouse knockout models for single chemokines [61]. In the human thymus, chemokines are differentially expressed by epithelial cells of the cortex, medulla, and CMJ in a pattern of expression similar to that in mouse [8, 20, 6265]. Chemokines may be tethered to the cell surface or secreted into the environment and bound by the ECM to establish a gradient [61]. Thymocytes, in turn, respond to chemokines via developmentally regulated expression of chemokine receptors, which helps thymocytes tune their chemotactic response to the local expression of ligands, and aids in migration from one microenvironment to the next (Table 2). Additionally, chemokines are important regulators of integrin avidity through the activation of inside-out signaling, which is implicated in the modulation of integrin adhesion [60].

Several other factors are known to modify thymocyte interactions with their microenvironment and may provide additional levels of modulation of thymocyte migration. These include galectins, cytokines, matrix metalloproteases, and glycosylaminoglycans. Additionally, semaphorins have recently been shown to modulate thymocyte interactions with the thymic microenvironment. Semaphorins bind to neuropilins, transmembrane glycoproteins classically known for their role in neuronal guidance during development, and their co-receptors, plexins, expressed by thymocytes, TECs, and thymic DCs. Semaphorins have been shown to modulate the adhesive capacity of neuropilin-expressing thymocytes and can also dampen chemokine induced migration [66, 67].

Progenitor entry into the thymus

T cell progenitors arise from hematopoietic stem cells in the BM and journey through the blood to provide the thymus with a continuous supply of progenitors to maintain thymopoiesis. Several BM-derived human thymic progenitor cells have been identified. CD34hiCD45+CD7+ hematopoietic progenitor cells (HPCs) are thought to be one of the earliest human thymus-seeding populations and retain the potential to differentiate into T, natural killer (NK), and B cells [68]. These HPCs emerge from the fetal BM at 8–9 weeks of gestation, and persist in significantly lower numbers into adult life [68]. Furthermore, CD34+CD7 progenitors with erythroid, myeloid, and lymphoid differentiation potential have been found in postnatal human thymi, indicating that CD7 multipotent progenitors may also contribute to human thymopoiesis [69]. Evidence that these cells are thymus-settling progenitors derives from the detection of these populations in both the BM and thymus. Additionally, it has been shown that BM CD34hiCD45+CD7+ hematopoietic cells are direct precursors of the earliest CD34hiCD1a fetal thymocytes and are attracted by, and have the ability to enter, the thymic parenchyma in ex vivo assays [68]. A separate group has further shown that in vitro-generated CD34hiCD45+CD7+ HPCs have the ability to colonize the thymus in a humanized mouse model [70]. Whether additional BM-derived cells contribute to the thymic progenitor population and how these populations vary throughout ontogeny is not known.

While progenitor seeding of the postnatal murine thymus occurs in a focal manner via the vasculature at the CMJ, there is evidence of multifocal seeding of the human and murine thymus prior to establishment of the vasculature during fetal development [20, 68, 71]. First, there is scattered cortical distribution of the earliest CD34+ immigrants in the human fetal thymus [20, 68]. In addition, it has been shown that human CD34hiCD45+CD7+ HPCs displayed a heightened ability to enter the thymic parenchyma in ex vivo assays that closely mimic the earliest stages of thymus colonization prior to thymic vascularization. In these studies, fluorescently labeled human CD34hiCD45+CD7+ HPCs seeded onto alymphoid thymic lobes from non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mouse embryos showed efficient and consistent infiltration into the thymic parenchyma across the basement membrane of the lobes and displayed a substantial capacity to generate TCRαβ+ DP thymocytes [68].

Chemokines appear to play an important role in progenitor seeding of the human thymus. In the avascular murine fetal thymus, both CCL25 and CCL21 are required for progenitor seeding of the tissue (Fig. 2a) [7275]. Similarly, the expression of the chemokine CCL25 as early as the 8th week of gestation in the human thymic parenchyma, along with expression of its receptor, CCR9, on BM CD34hiCD45+CD7+ cells, coincides with the appearance of the earliest HPCs within the human fetal thymus, suggesting a role for this chemokine in guiding human progenitor cells to the thymus (Fig. 2a) [20, 68]. Although expression of CCL21 is absent from the human fetal thymus until week 11, it is possible that, as in the mouse, CCL21 might instead be expressed by the adjacent parathyroid primordia, thus coordinating with thymic CCL25 expression to guide thymic progenitor entry into the avascular human fetal thymus [20, 75, 76]. Additionally, hematopoietic progenitors fail to enter the thymus of Foxn1nu (nude) mice, which may be accounted for by the absence of CCL25 expression by Foxn1-deficient thymic epithelial cells [72, 77, 78]. The common SCID phenotype in both mice and humans with Foxn1 deficiencies suggests highly conserved Foxn1 gene expression and function, further implicating chemokine signaling in progenitor entry into the human thymus.

Fig. 2.

Fig. 2

Comparative overview of the factors that direct thymocyte migration in mice and men. a Entry of hematopoietic progenitors into the vascularized murine thymus at the CMJ requires binding of P-selectin on thymic endothelial cells through its ligand PSGL-1, on thymus-seeding progenitor cells, as well as expression of CCL21 and CCL25 by the thymic parenchyma. These factors likely also mediate progenitor entry into the human thymus. b In the mouse, the outward migration of DN thymocytes towards the sub-capsular region of the thymus is mediated by CCL25 and CXCL12. The preserved cortical predominance of these two chemokines in humans suggests they may guide the migration of DN thymocytes through the human cortex. c DP thymocytes scan the cortex in search of positive selecting signals, and CXCL12 is required to maintain the cortical localization of human DP cells, while the retention of murine DP thymocytes in the cortex may be a chemokine-independent process. d Positively selected DP thymocytes that have received a low-affinity TCR signal differentiate to a SP stage and migrate to the medulla in response to CCL19/21 in both human and mouse. e Negative selection of auto-reactive T cells continues in the medulla, as well as maturation of SP thymocytes and differentiation of Treg cells. In humans, TSLP-conditioned dendritic cells are essential for Treg differentiation, and CCL22 may be involved in guiding Treg progenitors to the specialized niche surrounding the Hassall’s corpuscles. f In the mouse, mature SP thymocytes up-regulate S1P1 and exit the thymus along an S1P gradient to enter the circulation, and CXCL12 has additionally been implicated in this process in both mice and humans. There is data to suggest that S1P1 might also mediate the egress of human T cells from the thymus. Black text indicates factors that have been shown to influence thymocyte migration during development. Grey text indicates likely candidates to direct thymocyte migration that has not yet been proven. Blue dotted lines indicate dying cells. Asterisk indicates factors that appear to have unique, human-specific functions

The role of other chemokines in human progenitor settling of the thymus is less clear. Human BM-derived CD34+ progenitor cells with thymus-seeding potential also express CXCR4 and CCR7 and migrate in response to their respective ligands in vitro, suggesting a possible role for chemokine signaling in the recruitment and/or retention of human thymus colonizing cells during both fetal and adult stages of development [68, 7981]. However, studies examining the effect of CXCL12:CXCR4 inhibition on the ability of human CD34+ thymic precursors to repopulate murine fetal lobes have led to the suggestion that CXCR4 signaling is not required for progenitor entry into the thymus [7]. These studies used adult intrathymic progenitor cells that had presumably entered the thymus via the vasculature. Thus, it is important to also confirm these findings with early progenitor populations that are thought to seed the avascular fetal thymus. In addition, the ability of murine progenitor cells to settle the mature thymus is also contingent upon chemokine signaling through CCR9 and CCR7 [82, 83], but it is not yet known if these signals also influence human progenitor cell seeding of mature thymic tissue.

Entry of blood-borne lymphoid progenitors across the vascular bed of the mature human thymus likely mimics the requirements for entry into other tissues. Thus, it likely necessitates initiation of a rolling adhesion onto thymic microvasculature, followed by firm adhesion and cell arrest on vascular endothelium leading to extravasation of progenitors into the thymic parenchyma [60, 84]. In mice, it is well established that thymus-settling progenitors enter the adult thymic stroma at the well-perfused CMJ and that progenitor entry is dependent on the interaction between P-selectin on thymic endothelium and P-selectin glycoprotein ligand 1 (PSGL-1) on hematopoietic cells (Fig. 2a) [85, 86]. Data on the mechanism by which human progenitors enter the mature, vascularized thymus is scarce, likely due to the limited availability of models to address these questions. Assuming a certain degree of interspecies conservation, PSGL1 is expressed by early lineage marker negative (CD2, CD3, CD14, CD16, CD19, CD24, CD56, CD66b, and glycophorin-A) post-natal human thymocytes and to a lesser extent by in vitro generated CD34+CD7+CD5+ progenitor cells with thymus-colonizing potential, and thus may also direct human progenitor seeding of the thymus (Fig. 2a) [70]. Another potential mediator of human progenitor thymic entry is via an L-selectin:peripheral node addressin (PNAd) interaction, as these molecules are expressed on BM-derived CD34+CD7+ cells and by the endothelium of the human thymus, respectively [87, 88]. It is interesting that BM endothelial cell-derived CXCL12 plays a role in the initiation of rolling adhesion and cell arrest on endothelial receptors of the human BM such as E-selectin, P-selectin, intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), and it is possible a similar process may mediate progenitor entry across the thymic vasculature [89, 90].

In sum, there is emerging evidence that chemokines and adhesion molecules direct and mediate hematopoietic progenitor cells to enter and seed the human thymus. Although direct confirmatory evidence of the signaling cues involved in progenitor settling of the thymus is lacking in humans, certain findings suggest interspecies similarities. In addition to further studies on these topics, it will also be interesting to note the interplay between chemokines and adhesion molecules in these processes.

Progression of cell surface marker expression during human thymocyte development

Human thymus-colonizing progenitor cells retain multi-lineage differentiation potential, although the capacity to develop along the B cell lineage is likely lost once exposed to the thymic environment [68, 91, 92]. Commitment to the T cell lineage requires Notch signaling and is marked by up-regulation of CD1a expression on human CD34+ thymocytes [9194]. The most immature cells in the human thymus are a heterogeneous population of DN cells that can be further subdivided on the basis of CD34 and CD1a expression into consecutive stages of differentiation similar to those in mouse. Human DN thymocytes then lose CD34 expression and progress to a CD3CD4+CD8 intermediate single positive stage (CD4ISP), before developing into DP cells, and ultimately CD4+ and CD8+ mature SP T cells (Fig. 3) [95].

Fig. 3.

Fig. 3

Progression of cell surface marker expression during T cell development. Sequential view of the different phenotypes during mouse and human thymocyte development. Human DN thymocytes can be subdivided on the basis of CD34 and CD1a expression into maturational stages analogous to those in mouse. Transition from the DN to the DP stage proceeds through a CD4+ ISP in humans, and CD69 up-regulation marks the early stages of positive selection. The final stages of human SP thymocyte maturation can be defined by the relative expression of CD69, CD1a, CD27, and CD45RA

Human β-selection and initiation of TCRα rearrangement occurs at the CD34+CD38+CD1a+ stage, similar to the timing of TCR gene rearrangement in mouse [94]. After completion of TCRα gene rearrangement and expression of an αβ TCR, DP cells that engage in productive low-affinity interactions with self-peptide:MHC complexes (pMHC) on thymic epithelial cells proceed through positive selection, while thymocytes that engage in high affinity interactions with pMHC are deleted by negative selection. As in mouse, CD69 expression marks the early stages of positive selection during human T cell development (Fig. 3) [96].

Positively selected thymocytes down-regulate either CD4 or CD8 to differentiate into SP thymocytes, which then undergo further negative selection and maturation in the medulla. Human SP thymocytes can be further classified according to expression of CD27, down-regulation of CD1a, acquisition of CD45RA, and loss of CD69 during terminal differentiation (Fig. 3) [96, 97].

Migration within the thymus

Cortical migration of thymocytes

In mice, the majority of chemokines expressed in the thymus are more abundant in the medulla relative to the cortex. The exceptions are CCL25 and CXCL12, and the outward migration of mouse DN thymocytes from the point of entry at the CMJ toward the sub-capsular zone is mediated by cortical gradients to these chemokines, as well as CCL21 (Fig. 2b) [8, 86, 98101]. Conserved cortical-biased expression of CCL25 and CXCL12 point to a prominent role for CCR9 and CXCR4 signaling in guiding the migration of immature human cortical thymocytes (Fig. 2b) [9, 20]. Additionally, progressive differentiation of human DN thymocytes is accompanied by up-regulation of CCR7 and CCR9 surface expression, along with consistently high levels of CXCR4 expression [68, 102104]. Further, it has been shown that human DN cells migrate in vitro in response to CXCL12, CCL25, and modestly to CCL21, the same three chemokines that have been suggested to play a role in the cortical migration of murine DN thymocytes [16, 105].

In addition to their ability to attract thymocytes, chemokines have also been shown to influence cell adhesion through modulation of integrin activation [89, 90]. In particular, CXCL12 has been shown to activate the integrins VLA-4/α4β1 and VLA-5/α5β1 on immature human CD34+ hematopoietic cells. Interestingly, expression of CXCL12 is highest in the human cortex, and expression of its receptor, CXCR4, is highest among human DN thymocytes which also express high levels of both α4β1 and α5β1 and have been shown to adhere to fibronectin in vitro [9, 16, 106, 107]. Thus, it is likely that chemokines not only provide directional cues to developing thymocytes but also regulate thymocyte adhesion to the ECM, contributing to the distinct migration patterns of the different thymocyte populations [59].

Developmentally regulated expression of chemokine receptors continues at the DP stage of thymocyte differentiation, suggesting a role for chemokine signaling in the cortical localization of DP thymocytes. However, mice deficient for the receptors to CCL25 and CXCL12, the two exceptions to the predominantly medullary expression of chemokines, do not demonstrate overt defects at the DP stage of thymocyte development [73, 98, 99, 101, 108, 109]. Additionally, murine DP thymocytes overlaid on murine thymic slices were excluded from the medulla along a sharply defined boundary [110]. These observations have supported the notion that the cortical restriction of mouse DP thymocytes is a chemokine-independent process, and may instead be mediated by the inability of DP thymocytes to migrate on medullary substrate [110, 111]. In contrast, in vitro studies demonstrate that human pre-selection DP thymocytes (CD3loCD69) express high levels of CXCR4 and exhibit robust migration toward CXCL12 [16, 103, 112, 113]. Significantly, the transition through positive selection results in down-regulation of CXCR4 and loss of chemotaxis toward CXCL12, as well as improved migration to CCL21 coincident with CCR7 up-regulation in both humans and mice [16, 103, 112117]. However, chemotaxis towards another predominantly cortical chemokine, CCL25, did not differ between pre- and post-selection human DP thymocytes [16]. To determine the role of chemokines in directing the migration of human thymocytes in situ, we adapted a thymic slice model in which purified human thymocytes are overlaid on either human or mouse thymic slices. Using this system, we demonstrated that inhibition of CXCR4 signaling resulted in the striking loss of cortical localization of pre-selection DP thymocytes, which instead accumulated in the medulla [16]. Importantly, this finding held true on both murine and human thymic stroma and was independent of whether CXCR4 signaling was inhibited before or after DP thymocytes were allowed to migrate into the cortex. These results indicate that CXCL12 is required to both direct and retain human pre-selection DP thymocytes to the cortex (Fig. 2c).

A prominent role for chemokine signaling in the accumulation of human DP thymocytes to the cortex does not preclude the involvement of integrin-mediated adhesion in mediating localization of human DP thymocytes. In fact, given the role of chemokines in the modulation of integrin avidity for their ligands, integrins are likely key players in the modulation of thymocyte motility [60, 90]. Additionally, expression of integrins is developmentally regulated in human thymocytes, suggesting distinct roles during T cell maturation [106, 118, 119]. Constitutive activation of α4β1 in CD3loCD69 pre-selection DP thymocytes mediates firm adhesion to fibronectin in vitro and results in a stationary state, whereas CD3hiCD69+ post-selection DP thymocytes do not adhere to fibronectin despite equivalent levels of α4β1 surface expression. In fact, the interaction of CD3hiCD69hi DP thymocytes and mature SP thymocytes with fibronectin triggers migration rather than adhesion in a transwell assay, and involves ligation of both α4β1 and α5β1 [119].

Adding to the complexity of signaling interactions that are integrated by developing thymocytes to direct their migration in vivo, there is evidence for an additional level of control. TCR signaling has been demonstrated to induce expression of neuropilin-1 (NP-1) in human thymocytes, and ligation of its natural ligand, semaphorin-3A, on cortical and medullary TECs resulted in an inhibition of thymocyte adhesion [66]. More recent data demonstrates that semaphorin-3A additionally induces loss of chemotaxis towards CXCL12, and that this effect is mediated by down-regulation of CXCR4 expression among human DP and SP thymocytes [67]. These in vitro studies raise the interesting possibility that CXCR4 signaling, required for the cortical retention of human pre-selection DP thymocytes, may be modulated by TCR signaling during positive selection through the inhibitory effects of semaphorins. Chemokine signaling, in turn, affects integrin activation and thus modulates the ability of pre- and post-selection DP thymocytes to migrate on fibronectin. These findings suggest a tight integration of chemokine signaling, TCR activation, and adhesion to the ECM in the retention of human pre-selection DP thymocytes in the cortex.

Studies in the mouse have revealed a context-dependent effect of TCR signaling on thymocyte motility. We have noted that low-level TCR signals are required to sustain the motility of human polyclonal DP thymocytes consistent with indications of tonic TCR signaling in murine polyclonal DP thymocytes induced by MHC [16, 120]. This low-level tonic signaling is likely important for the generation of efficient positive selection signals, although the effect of altered motility on selection of the TCR repertoire and its influence on protective immunity has not been examined [53]. On the other hand, high-avidity TCR:MHC interactions associated with negative selection can lead to reduced motility and migratory arrest [53, 57, 121]. Using murine MHC class I- and II-deficient thymic slices in the chimeric human:mouse thymic slice model, we recently demonstrated that mouse MHC:human TCR signaling can support the activation and motility of polyclonal human DP thymocytes [16].

Both DN and DP murine thymocytes exhibit slow, confined migration patterns in the thymic cortex, and this behavior is conserved in immature human thymocytes. Two-photon imaging of human thymocyte migration within an intact thymic environment has revealed that DN and pre-selection DP thymocytes migrate with average speeds of 7 microns/min and ~4 microns/min, respectively [16]. These values are remarkably similar to those for mouse DN and DP thymocytes within the murine thymic cortex [120122]. However, how cortical chemokines, adhesion molecules, and other factors regulate cortical thymocyte speed is not known. In mouse models, it is known that positive selection of DP thymocytes correlates with significant increases in motility [122]. Although similar data is not available at present to confirm this observation in human thymocytes, it is tempting to speculate that the significant transition in chemokine receptor expression, chemokine sensitivity, integrin expression/activation, and TCR signaling contribute to these drastic changes in speed, and is consistent with the effects of some of these molecules on the in vitro migration of pre- and post-selection thymocytes. On the other hand, inhibition of chemokine signaling with pertussis toxin did not have a major impact on the overall motility of human pre-selection DP cells in situ [16]. However, the non-specific inhibition of all G-protein coupled receptors by pertussis toxin does not allow for a careful examination of the specific contribution of any one chemokine to motility, nor does it help discriminate between chemokines and other G-protein coupled receptors. Thus, the impact of chemokine signaling, integrin-mediated adhesion, and TCR signaling on the speed of immature human thymocytes requires further study.

Medullary migration of positively selected thymocytes

Prior to positive selection, high expression of CXCR4 allows the cortical predominance of CXCL12 to retain human DP thymocytes in the cortex. However, successful positive selection is accompanied by down-regulation of CXCR4 and up-regulation of CCR7, thereby promoting chemotaxis towards the medullary chemokines CCL19/21 [16, 62, 103, 112, 113]. The human medulla expresses a number of other chemokines, including MDC/CCL22, a ligand for CCR4 [63]. Interestingly, DP and SP human thymocytes also show enhanced chemotaxis towards CCL22, yet all CCR4+ thymocytes localize to the medulla [62], suggesting that some post-selection DP thymocytes may migrate to the medulla prior to differentiation into mature SP cells.

Mutations in CCR7 signaling in mice result in impaired SP thymocyte migration to the medulla and the development of autoimmunity, highlighting the importance of appropriate migration in shaping the TCR repertoire (Fig. 2d, e) [123, 124]. As in the mouse, migration toward CCL21 is also developmentally regulated in human thymocytes, with the greatest response occurring in the most mature CD1a SP cells [16, 62]. We recently examined the localization of human SP thymocytes on thymic slices from mice lacking CCR7 ligands (plt/plt mice), and found that CCR7 signaling is also necessary for the localization of mature human SP thymocytes to the medulla (Fig. 2d) [16]. A prominent role for CCR7 signaling in the medullary accumulation of human SP cells is not incompatible with a redundant role for CCL22, as the abnormal development of medullary epithelial cells in plt/plt mice may prevent the expression of CCL22, thus obscuring the contribution of CCL22 to the medullary localization of SP thymocytes.

In the human thymus, medullary epithelial cells of the Hassall’s corpuscles are thought to play a role in the generation of human regulatory T cells (Treg cells) through the production of thymic stromal lymphopoietin (TSLP). TSLP activates medullary DCs that mediate the selection of Treg cells [52]. Interestingly, medullary epithelial cells of the Hassall’s corpuscles also produce CCL22, and human Treg cells in the periphery express CCR4 and migrate in response to CCL22 [125]. Therefore, it is conceivable that CCR4 signaling could direct CCL22-responsive post-selection DP and immature SP human thymocytes to specialized niches of the medulla, such as those surrounding the Hassall’s corpuscles, and play a role in the generation of Treg cells (Fig. 2e).

Other chemokines produced primarily in the human thymic medulla include the ligands for CXCR3: IP-10 (CXCL10), Mig (CXCL9), and I-TAC (CXCL11), the majority of which are produced by epithelial cells [64]. CXCR3 expression is confined to mature human CD8+TCRαβ+ SP thymocytes, and CD3+TCRγδ+ cells. Chemotactic activity towards CXCL9, 10, and 11 is highest among mature human CD8+ SP thymocytes (CD45ROCD45RA+), but has no influence on CD4+ SP cells. CD3+TCRγδ+ T cells also migrate in response to these chemokines [64]. Human CD8+ SP and CD4+ SP thymocytes also migrate in response to CCL11/eotaxin, another medullary chemokine [65]. Whether these chemokines play a role in the medullary migration or localization of SP thymocytes in vivo is not yet known, but it is possible that thymocytes with distinct developmental potentials may be directed to defined medullary niches to finalize their maturation.

The migration of SP thymocytes in the medulla is associated with a striking increase in thymocyte motility, from the relatively slow migration of human DP thymocytes to average speeds of ~9 and 12 microns/min in CD4+ and CD8+ SP thymocytes respectively, similar to the speeds reported for mouse SP thymocytes [16, 110, 121, 126]. Remarkably, the speed of human and mouse SP thymocytes is maintained on both cortical and medullary substrates, suggesting that rapid migration is intrinsic to the developmental stage of the cell [16, 110]. Studies in mice have revealed that CCR7 signaling is additionally involved in determining the rapid motility of SP medullary thymocytes [110]. The contribution of G-protein coupled receptors, such as chemokines, to SP thymocyte motility was confirmed in humans, as treatment with pertussis toxin significantly reduced the average speed of SP thymocytes [16]. Thus, chemokine signaling, in combination with the developmentally regulated modulation of adhesion through integrin receptors, is likely responsible for the characteristic patterns of migration at different stages of thymocyte differentiation. Dissecting the contribution of individual integrins and the factors that control their activation merits further investigation and should yield important insight into the regulation of thymocyte migration both in the cortex and the medulla.

Exit from the thymus

Emigration of mature thymocytes into the circulation is required to maintain the peripheral naïve T cell pool and requires careful timing of thymic egress to ensure that thymocytes are effectively screened for reactivity to self-antigens in the medulla before they enter the circulation. In mice, emigration from the thymus is mediated by the regulated expression of S1P1, the receptor for the sphingolipid metabolite sphingosine-1-phosphate (S1P), on the most mature thymocytes (Fig. 2f) [127]. Lymphocyte egress from the thymus and from peripheral lymphoid organs is thought to occur in response to an S1P gradient, with low S1P levels in lymphoid tissue and high S1P levels in the circulation [128, 129], while the timing of thymocyte egress was shown to be tightly regulated by the effects of TCR signaling and CD69 expression on surface expression of S1P1 [129, 130]. Furthermore, transgenic expression of S1P1 on immature thymocytes led to their premature exit from the thymus, leading to insufficient negative selection and autoimmunity [129].

The role of S1P in T cell trafficking was initially revealed by the effect of FTY720 (fingolimod), an agonist/functional antagonist of S1P1 [131, 132]. FTY720 was consistently shown to prevent lymphocyte egress from secondary lymphoid tissue in mice, and this effect is thought to be mediated by the initial activation and eventual down-regulation and degradation of S1P1 [133]. Since then, FTY720 has been found to be effective in multiple animal models of autoimmune disease, particularly in experimental autoimmune encephalomyelitis, a model of human multiple sclerosis (MS) [133, 134]. Following promising clinical trials, FTY720 was approved for the treatment of relapsing MS. Similar to its effect on mouse lymphocytes, FTY720 treatment of MS patients was found to cause significant lymphopenia, specifically reducing the number of circulating naïve T cells and central memory T cells but not effector memory T cells [135]. Despite the absence of data regarding the effect of S1P modulation on human thymocyte egress, the conserved interspecies effect of FTY720 in peripheral T cell trafficking would suggest a prominent role for S1P in the exit of human lymphocytes from the thymus (Fig. 2f).

The blood vessels of the CMJ have been identified as the primary site of egress for the majority of murine thymocytes, while a role for lymphatic-mediated egress has been excluded [129, 136]. The absence of lymphatics in the human thymic medulla, as well as the presence of thymocytes within the perivascular spaces of human thymi, suggests that human thymocytes likely egress via blood vessels as well, although direct experimental evidence for this is lacking [129, 137, 138].

A role for chemokines in mediating thymic egress has also been proposed. Specifically, data from both FTOC and in vivo studies in mice demonstrated a role for CXCR4 signaling in the emigration of CD4+, but not CD8+, SP thymocytes (Fig. 2f) [139]. This suggests that murine CD4+ and CD8+ SP thymocytes may exit the thymus by two different mechanisms. In human thymocytes, however, the opposite appears to be true; both expression of CXCR4 and migration towards CXCL12 are greater in human CD8+ compared to CD4+ SP thymocytes [16]. Additionally, it has been previously demonstrated that both CD4+ and CD8+ human fetal SP thymocytes migrate away from thymic fragments and from a novel artificial thymic organoid (generated on a 3-D inert matrix) in a CXCL12-dependent manner, suggesting that human SP thymocyte egress is mediated by chemorepulsion to the chemokine CXCL12 produced by the thymic stroma (Fig. 2f) [140]. It will be interesting to determine the contribution of CXCL12 to thymocyte egress in the vascularized thymus of humanized mice that might more closely approximate conditions for thymic egress in vivo.

Chemokine-induced migration requires cell polarization that results from cytoskeleton remodeling. Coronin 1A, involved in actin cytoskeletal rearrangement, was found to be deficient in some patients with SCID, and coronin 1A-deficient mice have impaired thymic egress [141, 142]. Mammalian STE20-like protein kinase 1 (MST1), another molecule involved in the regulation of cytoskeletal remodeling, has been shown to be essential for murine thymic egress and lymphocyte migration [143]. It has therefore been suggested that the T cell lymphopenia observed in MST1-deficient patients might also be a consequence of impaired egress of mature T cells from the thymus [144].

Concluding remarks

Thymic T cell development is essential for the generation of protective immunity against foreign antigens, and the carefully orchestrated journey of thymocytes through the thymus is crucial to the correct development and maturation of T cells. Simultaneously, thymocyte signaling supports the differentiation of the thymic stroma, further underscoring the importance of thymocyte migration. Signaling interactions that guide the journey of thymocytes are complex and involve integration of a multitude of cues, some of which have been identified, and many others that are inferred from mouse studies. While a variety of in vitro systems and organ culture systems are available, the humanized mice model remains the only “in vivo” model for the dissection of human immune development. Despite significant advances, many aspects of human thymocyte migration, and their relation to T cell development, remain unknown.

Although candidate chemokines and adhesion molecules have been identified, studies of human thymic progenitor entry into and lymphocyte exit from the thymus are particularly hampered by the lack of models to efficiently recapitulate the trafficking of human cells between the intravascular space and the thymus. These studies are complicated by the fact that multiple different hematopoietic populations, some of which may not yet have been identified, may enter the thymus via unique mechanisms. Humanized mice are a promising experimental platform to study the signaling cues guiding the migration of human cells to and from the thymus, but certain limitations, such as inefficient interspecies receptor:ligand interactions and differences in the vascularization of the murine and human thymus, may complicate the effectiveness of these models. Despite the challenges, these questions remain an area of paramount importance, as the slow recovery of T cells following BM transplantation affects the morbidity and mortality of a large number of patients. Additionally, the current clinical use of FTY720, as well as the development of next generation S1P receptor modulators for the treatment of a wide range of inflammatory diseases, stresses the need for a closer characterization of the role of the S1P:S1P1 axis in the regulation of human thymocyte egress.

The thymic cortex harbors immature thymocytes as they progress through the early stages of T cell development, yet the signals directing the localization of human cortical thymocytes remains elusive. Assuming that the vessels of the CMJ are the site of hematopoietic cell entry into the vascularized human thymus as they are in mouse, it is still not known whether an outward migration to the sub-capsular region of the thymus is essential for progression through β-selection. Moreover, while CXCL12 signaling has been implicated in the retention of pre-selection DP thymocytes in the cortex, the identity of the signaling cues that guide the migration of human DN thymocytes in situ have yet to be identified. Additionally, integrins and adhesion molecules are likely implicated in the unique migration signatures of thymocytes and may influence the outcome at the various selection checkpoints in the thymic cortex. Thus, the contribution of integrins to thymocyte migration and development warrants further investigation.

The requirement for CCR7 signaling in the relocalization of post-selection thymocytes from the cortex to the medulla appears to be preserved between human and mouse. Whether some post-selection DP thymocytes migrate to the medulla prior to maturation to the SP stage, and whether other chemokine signals or alterations in thymocyte adhesion additionally contribute to the localization of immature SP thymocytes to the medulla is not known. It is likely that the human medulla contains specialized niches with distinct functional capacities, such as the contribution of Hassall’s corpuscles to the induction of human Treg cells. The medulla is rich in chemokine expression, and specific human thymocyte subsets have been shown to respond differentially to many of these in vitro. The physiologic relevance of directed thymocyte migration within specific niches of the medulla and its possible contribution to the selection of the TCR repertoire remains to be determined.

The ability to modulate thymocyte seeding of, migration within, and exit from the thymus is of great clinical interest in the treatment of disease. In addition to their role in thymocyte adhesion and migration, chemokines and integrins have been implicated in a significant number of pathologies, including autoimmune disorders, transplant rejection, cancer, and human immunodeficiency virus (HIV) infection. Thus, many small molecule antagonists targeting these receptor/ligand interactions are already in use, many others are in clinical trials, and the repertoire of targeted molecules is likely to continue to expand. Therefore, it is of paramount importance to understand the contribution of these molecules to thymocyte migration and development for the safe design of chemokine- and integrin- antagonists for clinical use.

Acknowledgments

We thank Brian Wiest, Claudia Brockmeyer, and Nadia Kurd for critical reading of the manuscript. This work was supported by the California Institute of Regenerative Medicine clinical fellowship TG2-01164 (to JH), post-doctoral training grant T1-00007 (to HJM), and grant RM1-01732 (to EAR).

Abbreviations

AIRE

Autoimmune regulator

BM

Bone marrow

CMJ

Cortical medullary junction

cTEC

Cortical thymic epithelial cell

DCs

Dendritic cells

DN

CD4CD8, double negative

DP

CD4+CD8+, double positive

ECM

Extracellular matrix

FTY720

Fingolimod

FTOC

Fetal thymic organ culture

HPCs

Hematopoietic progenitor cells

ICAM-1

Intercellular adhesion molecule 1

IL2Rgamma

IL2 receptor gamma chain

mTEC

Medullary thymic epithelial cell

MS

Multiple sclerosis

MST1

Mammalian sterile 20-like protein kinase 1

NOD

Non-obese diabetic

pMHC

Peptide-MHC complexes

PNAd

Peripheral node addressin

PSGL-1

P-selectin glycoprotein ligand 1

SCID

Severe combined immune deficiency

SP

CD4+CD8 or CD8+CD4, single positive

S1P

Sphingosine-1-phosphate

TCR

T cell receptor

TEC

Thymic epithelial cell

TSLP

Thymic stromal lymphopoietin

VCAM-1

Vascular cell adhesion molecule 1

References

  • 1.Nakagawa T, Roth W, Wong P, et al. Cathepsin L: critical role in li degradation and CD4 T cell selection in the thymus. Science. 1998;280:450–453. doi: 10.1126/science.280.5362.450. [DOI] [PubMed] [Google Scholar]
  • 2.Murata S, Sasaki K, Kishimoto T, et al. Regulation of CD8+ T cell development by thymus-specific proteasomes. Science. 2007;316:1349–1353. doi: 10.1126/science.1141915. [DOI] [PubMed] [Google Scholar]
  • 3.Honey K, Nakagawa T, Peters C, Rudensky A. Cathepsin L regulates CD4+ T cell selection independently of its effect on invariant chain: a role in the generation of positively selecting peptide ligands. J Exp Med. 2002;195:1349–1358. doi: 10.1084/jem.20011904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.McCaughtry TM, Baldwin TA, Wilken MS, Hogquist KA. Clonal deletion of thymocytes can occur in the cortex with no involvement of the medulla. J Exp Med. 2008;205:2575–2584. doi: 10.1084/jem.20080866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Anderson MS, Venanzi ES, Klein L, et al. Projection of an immunological self shadow within the thymus by the AIRE protein. Science. 2002;298:1395–1401. doi: 10.1126/science.1075958. [DOI] [PubMed] [Google Scholar]
  • 6.Annunziato F, Romagnani P, Cosmi L. Chemokines and lymphopoiesis in human thymus. Trends Immunol. 2001;22:277–281. doi: 10.1016/s1471-4906(01)01889-0. [DOI] [PubMed] [Google Scholar]
  • 7.Hernández-López C, Varas A. Stromal cell-derived factor 1/CXCR4 signaling is critical for early human T-cell development. Blood. 2002;99:546–554. doi: 10.1182/blood.v99.2.546. [DOI] [PubMed] [Google Scholar]
  • 8.Griffith AV, Fallahi M, Nakase H, et al. Spatial mapping of thymic stromal microenvironments reveals unique features influencing T lymphoid differentiation. Immunity. 2009;31:999–1009. doi: 10.1016/j.immuni.2009.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gotter J, Brors B, Hergenhahn M, et al. Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters. J Exp Med. 2004;199:155–166. doi: 10.1084/jem.20031677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Entschladen F, Drell TL, IV, Lang K, et al. Analysis methods of human cell migration. Exp Cell Res. 2005;307:418–426. doi: 10.1016/j.yexcr.2005.03.029. [DOI] [PubMed] [Google Scholar]
  • 11.Weber M, Hauschild R, Schwarz J, et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science. 2013;339:328–332. doi: 10.1126/science.1228456. [DOI] [PubMed] [Google Scholar]
  • 12.Caserta S, Campanello S, Tomaiuolo G, Sabetta L, Guido S. A Methodology to study chemotaxis in 3-D collagen gels. AIChE J. 2013;59:4025–4035. [Google Scholar]
  • 13.Plum J, Smedt M, Leclercq G, et al. Human intrathymic development: a selective approach. Semin Immunopathol. 2008;30:411–423. doi: 10.1007/s00281-008-0135-2. [DOI] [PubMed] [Google Scholar]
  • 14.Galy A, Verma S, Barcena A, Spits H. Precursors of CD3+CD4+CD8+ cells in the human thymus are defined by expression of CD34. Delineation of early events in human thymic development. J Exp Med. 1993;178:391–401. doi: 10.1084/jem.178.2.391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barcena A, Galy AH, Punnonen J, et al. Lymphoid and myeloid differentiation of fetal liver CD34+ lineage-cells in human thymic organ culture. J Exp Med. 1994;180:123–132. doi: 10.1084/jem.180.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Halkias J, Melichar HJ, Taylor KT, et al. Opposing chemokine gradients control human thymocyte migration in situ. J Clin Invest. 2013;123:2131–2142. doi: 10.1172/JCI67175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen Q, Khoury M, Chen J. Expression of human cytokines dramatically improves reconstitution of specific human-blood lineage cells in humanized mice. Proc Natl Acad Sci USA. 2009;106:21783–21788. doi: 10.1073/pnas.0912274106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Huntington ND, Alves NL, Legrand N, et al. Autonomous and extrinsic regulation of thymopoiesis inhuman immune system (HIS) mice. Eur J Immunol. 2011;41:2883–2893. doi: 10.1002/eji.201141586. [DOI] [PubMed] [Google Scholar]
  • 19.Eisenman J, Ahdieh M, Beers C, et al. Interleukin-15 interactions with interleukin-15 receptor complexes: characterization and species specificity. Cytokine. 2002;20:121–129. doi: 10.1006/cyto.2002.1989. [DOI] [PubMed] [Google Scholar]
  • 20.Farley AM, Morris LX, Vroegindeweij E, et al. Dynamics of thymus organogenesis and colonization in early human development. Development. 2013;140:2015–2026. doi: 10.1242/dev.087320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gordon J, Manley NR. Mechanisms of thymus organogenesis and morphogenesis. Development. 2011;138:3865–3878. doi: 10.1242/dev.059998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lobach DF, Haynes BF. Ontogeny of the human thymus during fetal development. J Clin Immunol. 1987;7:81–97. doi: 10.1007/BF00916002. [DOI] [PubMed] [Google Scholar]
  • 23.Haynes BFB, Heinly CSC. Early human T cell development: analysis of the human thymus at the time of initial entry of hematopoietic stem cells into the fetal thymic microenvironment. J Exp Med. 1995;181:1445–1458. doi: 10.1084/jem.181.4.1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Haynes BFB, Scearce RMR, Lobach DFD, Hensley LLL. Phenotypic characterization and ontogeny of mesodermal-derived and endocrine epithelial components of the human thymic microenvironment. J Exp Med. 1984;159:1149–1168. doi: 10.1084/jem.159.4.1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pignata C, Gaetaniello L, Masci AM, et al. Human equivalent of the mouse Nude/SCID phenotype: long-term evaluation of immunologic reconstitution after bone marrow transplantation. Blood. 2001;97:880–885. doi: 10.1182/blood.v97.4.880. [DOI] [PubMed] [Google Scholar]
  • 26.Nehls MM, Pfeifer DD, Schorpp MM, et al. New member of the winged-helix protein family disrupted in mouse and rat nude mutations. Nature. 1994;372:103–107. doi: 10.1038/372103a0. [DOI] [PubMed] [Google Scholar]
  • 27.Frank J, Pignata C, Panteleyev AA, et al. Exposing the human nude phenotype. Nature. 1999;398:473–474. doi: 10.1038/18997. [DOI] [PubMed] [Google Scholar]
  • 28.Finnish-German APECED Consortium An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet. 1997;17:399–403. doi: 10.1038/ng1297-399. [DOI] [PubMed] [Google Scholar]
  • 29.Nagamine K, Peterson P, Scott HS, et al. Positional cloning of the APECED gene. Nat Genet. 1997;17:393–398. doi: 10.1038/ng1297-393. [DOI] [PubMed] [Google Scholar]
  • 30.Havran WLW, Allison JPJ. Developmentally ordered appearance of thymocytes expressing different T-cell antigen receptors. Nature. 1988;335:443–445. doi: 10.1038/335443a0. [DOI] [PubMed] [Google Scholar]
  • 31.Ikuta K, Kina T, MacNeil I, et al. A developmental switch in thymic lymphocyte maturation potential occurs at the level of hematopoietic stem cells. Cell. 1990;62:863–874. doi: 10.1016/0092-8674(90)90262-d. [DOI] [PubMed] [Google Scholar]
  • 32.Spear PG, Wang AL, Rutishauser U, Edelman GM. Characterization of splenic lymphoid cells in fetal and newborn mice. J Exp Med. 1973;138:557–573. doi: 10.1084/jem.138.3.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stites DP, Pavia CS. Ontogeny of human T cells. Pediatrics. 1979;64:795–802. [PubMed] [Google Scholar]
  • 34.Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med. 1996;184:387–396. doi: 10.1084/jem.184.2.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cupedo T, Nagasawa M, Weijer K, et al. Development and activation of regulatory T cells in the human fetus. Eur J Immunol. 2005;35:383–390. doi: 10.1002/eji.200425763. [DOI] [PubMed] [Google Scholar]
  • 36.Darrasse-Jeze G, Marodon G, Salomon BL, Catala M, Klatzmann D. Ontogeny of CD4+CD25+ regulatory/suppressor T cells in human fetuses. Blood. 2005;105:4715–4721. doi: 10.1182/blood-2004-10-4051. [DOI] [PubMed] [Google Scholar]
  • 37.Michaëlsson J, Mold JE, McCune JM, Nixon DF. Regulation of T cell responses in the developing human fetus. J Immunol. 2006;176:5741–5748. doi: 10.4049/jimmunol.176.10.5741. [DOI] [PubMed] [Google Scholar]
  • 38.Holländer GAG, Wang BB, Nichogiannopoulou AA, et al. Developmental control point in induction of thymic cortex regulated by a subpopulation of prothymocytes. Nature. 1995;373:350–353. doi: 10.1038/373350a0. [DOI] [PubMed] [Google Scholar]
  • 39.Shores EWE, van Ewijk WW, Singer AA. Disorganization and restoration of thymic medullary epithelial cells in T cell receptor-negative scid mice: evidence that receptor-bearing lymphocytes influence maturation of the thymic microenvironment. Eur J Immunol. 1991;21:1657–1661. doi: 10.1002/eji.1830210711. [DOI] [PubMed] [Google Scholar]
  • 40.Klug DBD, Carter CC, Crouch EE, et al. Interdependence of cortical thymic epithelial cell differentiation and T-lineage commitment. Proc Natl Acad Sci USA. 1998;95:11822–11827. doi: 10.1073/pnas.95.20.11822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.van Ewijk W, Holländer G, Terhorst C, Wang B. Stepwise development of thymic microenvironments in vivo is regulated by thymocyte subsets. Development. 2000;127:1583–1591. doi: 10.1242/dev.127.8.1583. [DOI] [PubMed] [Google Scholar]
  • 42.Irla M, Hugues S, Gill J, et al. Autoantigen-specific interactions with CD4+ thymocytes control mature medullary thymic epithelial cell cellularity. Immunity. 2008;29:451–463. doi: 10.1016/j.immuni.2008.08.007. [DOI] [PubMed] [Google Scholar]
  • 43.Hikosaka Y, Nitta T, Ohigashi I, et al. The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator. Immunity. 2008;29:438–450. doi: 10.1016/j.immuni.2008.06.018. [DOI] [PubMed] [Google Scholar]
  • 44.Poliani PL, Facchetti F, Ravanini M, et al. Early defects in human T-cell development severely affect distribution and maturation of thymic stromal cells: possible implications for the pathophysiology of Omenn syndrome. Blood. 2009;114:105–108. doi: 10.1182/blood-2009-03-211029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Villa A, Notarangelo LD, Chaim M, Roifman MDF. Omenn syndrome: inflammation in leaky severe combined immunodeficiency. J Allergy Clin Immunol. 2008;122:1082–1086. doi: 10.1016/j.jaci.2008.09.037. [DOI] [PubMed] [Google Scholar]
  • 46.Villa AA, Santagata SS, Bozzi FF, et al. Partial V(D)J recombination activity leads to Omenn syndrome. Cell. 1998;93:12. doi: 10.1016/s0092-8674(00)81448-8. [DOI] [PubMed] [Google Scholar]
  • 47.Fischer A. Human primary immunodeficiency diseases. Immunity. 2007;27:835–845. doi: 10.1016/j.immuni.2007.11.012. [DOI] [PubMed] [Google Scholar]
  • 48.Cavadini P, Vermi W, Facchetti F, et al. AIRE deficiency in thymus of 2 patients with Omenn syndrome. J Clin Invest. 2005;115:728–732. doi: 10.1172/JCI23087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Marrella V, Poliani PL, Casati A, et al. A hypomorphic R229Q Rag2 mouse mutant recapitulates human Omenn syndrome. J Clin Invest. 2007;117:1260–1269. doi: 10.1172/JCI30928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Patel DD, Gooding ME, Parrott RE, et al. Thymic function after hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency. N Engl J Med. 2000;342:1325–1332. doi: 10.1056/NEJM200005043421804. [DOI] [PubMed] [Google Scholar]
  • 51.Sarzotti M, Patel DD, Li X, et al. T cell repertoire development in humans with SCID after nonablative allogeneic marrow transplantation. J Immunol. 2003;170:2711–2718. doi: 10.4049/jimmunol.170.5.2711. [DOI] [PubMed] [Google Scholar]
  • 52.Watanabe N, Wang Y-H, Lee HK, et al. Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+regulatory T cells in human thymus. Nature. 2005;436:1181–1185. doi: 10.1038/nature03886. [DOI] [PubMed] [Google Scholar]
  • 53.Melichar HJ, Ross JO, Herzmark P, et al. Distinct temporal patterns of T cell receptor signaling during positive versus negative selection in situ. Sci Signal. 2013;6:ra92. doi: 10.1126/scisignal.2004400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Volkmann A, Zal T, Stockinger B. Antigen presenting cells in the thymus that can negatively select MHC class II-restricted T cells recognizing a circulating self antigen. Immunol Lett. 1997;56:87–88. [PubMed] [Google Scholar]
  • 55.Surh CD, Sprent J. T-cell apoptosis detected in situ during positive and negative selection in the thymus. Nature. 1994;372:100–103. doi: 10.1038/372100a0. [DOI] [PubMed] [Google Scholar]
  • 56.Fadok VA, Voelker DR, Campbell PA, et al. Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J Immunol. 1992;148:2207–2216. [PubMed] [Google Scholar]
  • 57.Dzhagalov IL, Chen KG, Herzmark P, Robey EA. Elimination of self-reactive T cells in the thymus: a timeline for negative selection. PLoS Biol. 2013;11:e1001566. doi: 10.1371/journal.pbio.1001566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gameiro J, Nagib P, Verinaud L. The thymus microenvironment in regulating thymocyte differentiation. Cell Adhesion Migr. 2010;4:382–390. doi: 10.4161/cam.4.3.11789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Savino W. Molecular mechanisms governing thymocyte migration: combined role of chemokines and extracellular matrix. J Leukoc Biol. 2004;75:951–961. doi: 10.1189/jlb.1003455. [DOI] [PubMed] [Google Scholar]
  • 60.Kinashi T. Intracellular signalling controlling integrin activation in lymphocytes. Nat Rev Immunol. 2005;5:546–559. doi: 10.1038/nri1646. [DOI] [PubMed] [Google Scholar]
  • 61.Olson TS, Ley K. Chemokines and chemokine receptors in leukocyte trafficking. Am J Physiol Regul Integr Comp Physiol. 2002;283:R7–R28. doi: 10.1152/ajpregu.00738.2001. [DOI] [PubMed] [Google Scholar]
  • 62.Annunziato F, Romagnani P, Cosmi L, et al. Macrophage-derived chemokine and EBI1-ligand chemokine attract human thymocytes in different stages of development and are produced by distinct subsets of medullary epithelial cells: possible implications for negative selection. J Immunol. 2000;4:135–137. doi: 10.4049/jimmunol.165.1.238. [DOI] [PubMed] [Google Scholar]
  • 63.Chantry D, Romagnani P, Raport CJ, et al. Macrophage-derived chemokine is localized to thymic medullary epithelial cells and is a chemoattractant for CD3(+), CD4(+), CD8(low) thymocytes. Blood. 1999;94:1890–1898. [PubMed] [Google Scholar]
  • 64.Romagnani P, Annunziato F, Lazzeri E, et al. Interferon-inducible protein 10, monokine induced by interferon gamma, and interferon-inducible T-cell alpha chemoattractant are produced by thymic epithelial cells and attract T-cell receptor (TCR) alphabeta+ CD8+ single-positive T cells, TCRgammadelta+ T cells, and natural killer-type cells in human thymus. Blood. 2001;97:601–607. doi: 10.1182/blood.v97.3.601. [DOI] [PubMed] [Google Scholar]
  • 65.Franz-Bacon K, Dairaghi DJ, Boehme SA, et al. Human thymocytes express CCR-3 and are activated by eotaxin. Blood. 1999;93:3233–3240. [PubMed] [Google Scholar]
  • 66.Lepelletier YY, Smaniotto SS, Hadj-Slimane RR, et al. Control of human thymocyte migration by neuropilin-1/semaphorin-3A-mediated interactions. Proc Natl Acad Sci USA. 2007;104:5545–5550. doi: 10.1073/pnas.0700705104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Garcia F, Lepelletier Y, Smaniotto S, et al. Inhibitory effect of semaphorin-3A, a known axon guidance molecule, in the human thymocyte migration induced by CXCL12. J Leukoc Biol. 2011 doi: 10.1189/jlb.0111031. [DOI] [PubMed] [Google Scholar]
  • 68.Haddad R, Guimiot F, Six E, et al. Dynamics of thymus-colonizing cells during human development. Immunity. 2006;24:217–230. doi: 10.1016/j.immuni.2006.01.008. [DOI] [PubMed] [Google Scholar]
  • 69.Hao QL, George AA, Zhu J, et al. Human intrathymic lineage commitment is marked by differential CD7 expression: identification of CD7-lympho-myeloid thymic progenitors. Blood. 2007;111:1318–1326. doi: 10.1182/blood-2007-08-106294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Awong G, Herer E, Surh CD, et al. Characterization in vitro and engraftment potential in vivo of human progenitor T cells generated from hematopoietic stem cells. Blood. 2009;114:972–982. doi: 10.1182/blood-2008-10-187013. [DOI] [PubMed] [Google Scholar]
  • 71.Kyewski BAB. Seeding of thymic microenvironments defined by distinct thymocyte-stromal cell interactions is developmentally controlled. J Exp Med. 1987;166:520–538. doi: 10.1084/jem.166.2.520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bleul CC, Boehm T. Chemokines define distinct microenvironments in the developing thymus. Eur J Immunol. 2000;30:3371–3379. doi: 10.1002/1521-4141(2000012)30:12<3371::AID-IMMU3371>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 73.Wurbel MA, Malissen M, Guy-Grand D, Meffre E, Nussenzweig MC, Richelme M, Carrier A, Malissen B. Mice lacking the CCR9 CC-chemokine receptor show a mild impairment of early T- and B-cell development and a reduction in T-cell receptor gamma delta + gut intraepithelial lymphocytes. Blood. 2001;98:2626–2632. doi: 10.1182/blood.v98.9.2626. [DOI] [PubMed] [Google Scholar]
  • 74.Liu C, Ueno T, Kuse S, Saito F, Nitta T, Piali L, Nakano H, Kakiuchi T, Lipp M, Hollander GA, Takahama Y. The role of CCL21 in recruitment of T-precursor cells to fetal thymi. Blood. 2005;105:31–39. doi: 10.1182/blood-2004-04-1369. [DOI] [PubMed] [Google Scholar]
  • 75.Liu C, Saito F, Liu Z, et al. Coordination between CCR7- and CCR9-mediated chemokine signals in prevascular fetal thymus colonization. Blood. 2006;108:2531–2539. doi: 10.1182/blood-2006-05-024190. [DOI] [PubMed] [Google Scholar]
  • 76.Bleul CC, Fuhlbrigge RC, Casasnovas JM, et al. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1) J Exp Med. 1996;184:1101–1109. doi: 10.1084/jem.184.3.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Itoi M, Tsukamoto N, Amagai T. Expression of Dll4 and CCL25 in Foxn1-negative epithelial cells in the post-natal thymus. Int Immunol. 2006;19:127–132. doi: 10.1093/intimm/dxl129. [DOI] [PubMed] [Google Scholar]
  • 78.Holländer G, Gill J, Zuklys S, et al. Cellular and molecular events during early thymus development. Immunol Rev. 2006;209:28–46. doi: 10.1111/j.0105-2896.2006.00357.x. [DOI] [PubMed] [Google Scholar]
  • 79.Aiuti A, Webb IJ, Bleul C, et al. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185:111–120. doi: 10.1084/jem.185.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kim CH, Broxmeyer HE. SLC/exodus2/6Ckine/TCA4 induces chemotaxis of hematopoietic progenitor cells: differential activity of ligands of CCR7, CXCR3, or CXCR4 in chemotaxis vs. suppression of progenitor proliferation. J Leukoc Biol. 1999;66:455–461. doi: 10.1002/jlb.66.3.455. [DOI] [PubMed] [Google Scholar]
  • 81.Rosu-Myles M, Khandaker M, Wu DM, et al. Characterization of chemokine receptors expressed in primitive blood cells during human hematopoietic ontogeny. Stem Cells. 2000;18:374–381. doi: 10.1634/stemcells.18-5-374. [DOI] [PubMed] [Google Scholar]
  • 82.Zlotoff DA, Sambandam A, Logan TD, et al. CCR7 and CCR9 together recruit hematopoietic progenitors to the adult thymus. Blood. 2010;115:1897–1905. doi: 10.1182/blood-2009-08-237784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Krueger A, Willenzon S, Lyszkiewicz M, et al. CC chemokine receptor 7 and 9 double-deficient hematopoietic progenitors are severely impaired in seeding the adult thymus. Blood. 2010;115:1906–1912. doi: 10.1182/blood-2009-07-235721. [DOI] [PubMed] [Google Scholar]
  • 84.Scimone ML, Aifantis I, Apostolou I, et al. A multistep adhesion cascade for lymphoid progenitor cell homing to the thymus. Proc Natl Acad Sci USA. 2006;103:7006–7011. doi: 10.1073/pnas.0602024103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rossi FMV, Corbel SY, Merzaban JS, et al. Recruitment of adult thymic progenitors is regulated by P-selectin and its ligand PSGL-1. Nat Immunol. 2005;6:626–634. doi: 10.1038/ni1203. [DOI] [PubMed] [Google Scholar]
  • 86.Lind EFE, Prockop SES, Porritt HEH, Petrie HTH. Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. J Exp Med. 2001;194:127–134. doi: 10.1084/jem.194.2.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Terstappen LWL, Huang SS, Picker LJL. Flow cytometric assessment of human T-cell differentiation in thymus and bone marrow. Blood. 1992;79:666–677. [PubMed] [Google Scholar]
  • 88.Flores KG, Li J, Sempowski GD, et al. Analysis of the human thymic perivascular space during aging. J Clin Invest. 1999;104:1031–1039. doi: 10.1172/JCI7558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Peled A, Grabovsky V, Habler L, et al. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34+ cells on vascular endothelium under shear flow. J Clin Invest. 1999;104:1199–1211. doi: 10.1172/JCI7615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Peled A, Kollet O, Ponomaryov T, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000;95:3289–3296. [PubMed] [Google Scholar]
  • 91.De Smedt M, Hoeboke I, Reynvoet K, et al. Different thresholds of notch signaling bias human precursor cells toward B-, NK-, monocytic/dendritic-, or T-cell lineage in thymus microenvironment. Blood. 2005;106:3498–3506. doi: 10.1182/blood-2005-02-0496. [DOI] [PubMed] [Google Scholar]
  • 92.Weerkamp F, Baert MRM, Brugman MH, et al. Human thymus contains multipotent progenitors with T/B lymphoid, myeloid, and erythroid lineage potential. Blood. 2006;107:3131–3137. doi: 10.1182/blood-2005-08-3412. [DOI] [PubMed] [Google Scholar]
  • 93.De Smedt M, Reynvoet K, Kerre T, et al. Active form of notch imposes T cell fate in human progenitor cells. J Immunol. 2002;169:3021–3029. doi: 10.4049/jimmunol.169.6.3021. [DOI] [PubMed] [Google Scholar]
  • 94.Dik WA, Pike-Overzet K, Weerkamp F, et al. New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling. J Exp Med. 2005;201:1715–1723. doi: 10.1084/jem.20042524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kraft DL, Weissman IL, Waller EK. Differentiation of CD3-4-8-human fetal thymocytes in vivo: characterization of a CD3-4+8-intermediate. J Exp Med. 1993;178:265–277. doi: 10.1084/jem.178.1.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Vanhecke D, Verhasselt B, De Smedt M, et al. Human thymocytes become lineage committed at an early postselection CD69+ stage, before the onset of functional maturation. J Immunol. 1997;159:5973–5983. [PubMed] [Google Scholar]
  • 97.Vanhecke D, Leclercq G, Plum J, Vandekerckhove B. Characterization of distinct stages during the differentiation of human CD69+CD3+ thymocytes and identification of thymic emigrants. J Immunol. 1995;155:1862–1872. [PubMed] [Google Scholar]
  • 98.Plotkin J, Prockop SE, Lepique A, Petrie HT. Critical role for CXCR4 signaling in progenitor localization and T cell differentiation in the postnatal thymus. J Immunol. 2003;171:4521–4527. doi: 10.4049/jimmunol.171.9.4521. [DOI] [PubMed] [Google Scholar]
  • 99.Benz C, Heinzel K, Bleul CC. Homing of immature thymocytes to the subcapsular microenvironment within the thymus is not an absolute requirement for T? Cell development. Eur J Immunol. 2004;34:3652–3663. doi: 10.1002/eji.200425248. [DOI] [PubMed] [Google Scholar]
  • 100.Misslitz AA, Pabst OO, Hintzen GG, et al. Thymic T cell development and progenitor localization depend on CCR7. J Exp Med. 2004;200:481–491. doi: 10.1084/jem.20040383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Trampont PC, Tosello-Trampont A-C, Shen Y, et al. CXCR4 acts as a costimulator during thymic beta-selection. Nat Immunol. 2010;11:162–170. doi: 10.1038/ni.1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Aiuti A, Tavian M, Cipponi A, et al. Expression of CXCR4, the receptor for stromal cell-derived factor-1 on fetal and adult human lympho-hematopoietic progenitors. Eur J Immunol. 1999;29:1823–1831. doi: 10.1002/(SICI)1521-4141(199906)29:06<1823::AID-IMMU1823>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 103.Berkowitz RD, Alexander S, Bare C, et al. CCR5-and CXCR4-utilizing strains of human immunodeficiency virus type 1 exhibit differential tropism and pathogenesis in vivo. J Virol. 1998;72:10108–10117. doi: 10.1128/jvi.72.12.10108-10117.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zabel BA, Agace WW, Campbell JJ, et al. Human G protein-coupled receptor GPR-9-6/CC chemokine receptor 9 is selectively expressed on intestinal homing T lymphocytes, mucosal lymphocytes, and thymocytes and is required for thymus-expressed chemokine-mediated chemotaxis. J Exp Med. 1999;190:1241–1256. doi: 10.1084/jem.190.9.1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Youn BS, Kim CH, Smith FO, Broxmeyer HE. TECK, an efficacious chemoattractant for human thymocytes, uses GPR-9-6/CCR9 as a specific receptor. Blood. 1999;94:2533–2536. [PubMed] [Google Scholar]
  • 106.Mojcik CF, Salomon DR, Chang AC, Shevach EM. Differential expression of integrins on human thymocyte subpopulations. Blood. 1995;86:4206–4217. [PubMed] [Google Scholar]
  • 107.Gares SLS, Giannakopoulos NN, MacNeil DD, et al. During human thymic development, beta 1 integrins regulate adhesion, motility, and the outcome of RHAMM/hyaluronan engagement. J Leukoc Biol. 1998;64:781–790. doi: 10.1002/jlb.64.6.781. [DOI] [PubMed] [Google Scholar]
  • 108.Uehara S, Grinberg A, Farber JM, Love PE. A role for CCR9 in T lymphocyte development and migration. J Immunol. 2002;168:2811–2819. doi: 10.4049/jimmunol.168.6.2811. [DOI] [PubMed] [Google Scholar]
  • 109.Janas ML, Varano G, Gudmundsson K, et al. Thymic development beyond beta-selection requires phosphatidylinositol 3-kinase activation by CXCR4. J Exp Med. 2010;207:247–261. doi: 10.1084/jem.20091430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ehrlich LIR, Oh DY, Weissman IL, Lewis RS. Differential contribution of chemotaxis and substrate restriction to segregation of immature and mature thymocytes. Immunity. 2009;31:986–998. doi: 10.1016/j.immuni.2009.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Petrie HT. Cell migration and the control of post-natal T-cell lymphopoiesis in the thymus. Nat Rev Immunol. 2003;3:859–866. doi: 10.1038/nri1223. [DOI] [PubMed] [Google Scholar]
  • 112.Kitchen SG, Zack JA. CXCR4 expression during lymphopoiesis: implications for human immunodeficiency virus type 1 infection of the thymus. J Virol. 1997;71:6928–6934. doi: 10.1128/jvi.71.9.6928-6934.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Swainson L, Kinet S, Manel N, et al. Glucose transporter 1 expression identifies a population of cycling CD4+CD8+ human thymocytes with high CXCR4-induced chemotaxis. Proc Natl Acad Sci USA. 2005;102:12867–12872. doi: 10.1073/pnas.0503603102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Suzuki GG, Nakata YY, Dan YY, et al. Loss of SDF-1 receptor expression during positive selection in the thymus. Int Immunol. 1998;10:1049–1056. doi: 10.1093/intimm/10.8.1049. [DOI] [PubMed] [Google Scholar]
  • 115.Ueno T, Saito F, Gray DHD, et al. CCR7 signals are essential for cortex-medulla migration of developing thymocytes. J Exp Med. 2004;200:493–505. doi: 10.1084/jem.20040643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Campbell JJ, Pan J, Butcher EC. Cutting edge: developmental switches in chemokine responses during T cell maturation. J Immunol. 1999;163:2353–2357. [PubMed] [Google Scholar]
  • 117.Kim CH, Pelus LM, White JR, Broxmeyer HE. Differential chemotactic behavior of developing T cells in response to thymic chemokines. Blood. 1998;91:4434–4443. [PubMed] [Google Scholar]
  • 118.Salomon DR, Mojcik CF, Chang AC, et al. Constitutive activation of integrin alpha 4 beta 1 defines a unique stage of human thymocyte development. J Exp Med. 1994;179:1573–1584. doi: 10.1084/jem.179.5.1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Crisa LL, Cirulli VV, Ellisman MHM, et al. Cell adhesion and migration are regulated at distinct stages of thymic T cell development: the roles of fibronectin, VLA4, and VLA5. J Exp Med. 1996;184:215–228. doi: 10.1084/jem.184.1.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ladi E, Schwickert TA, Chtanova T, et al. Thymocyte-dendritic cell interactions near sources of CCR7 ligands in the thymic cortex. J Immunol. 2008;181:7014–7023. doi: 10.4049/jimmunol.181.10.7014. [DOI] [PubMed] [Google Scholar]
  • 121.Le Borgne M, Ladi E, Dzhagalov I, et al. The impact of negative selection on thymocyte migration in the medulla. Nat Immunol. 2009;10:823–830. doi: 10.1038/ni.1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Witt CM, Raychaudhuri S, Schaefer B, et al. Directed migration of positively selected thymocytes visualized in real time. PLoS Biol. 2005;3:e160. doi: 10.1371/journal.pbio.0030160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Nitta T, Nitta S, Lei Y, et al. CCR7-mediated migration of developing thymocytes to the medulla is essential for negative selection to tissue-restricted antigens. Proc Natl Acad Sci USA. 2009;106:17129–17133. doi: 10.1073/pnas.0906956106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kurobe H, Liu C, Ueno T, et al. CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity. 2006;24:165–177. doi: 10.1016/j.immuni.2005.12.011. [DOI] [PubMed] [Google Scholar]
  • 125.Iellem A, Mariani M, Lang R, et al. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4+CD25+ regulatory T cells. J Exp Med. 2001;194:847–854. doi: 10.1084/jem.194.6.847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bousso P, Robey EA. Dynamic behavior of T cells and thymocytes in lymphoid organs as revealed by two-photon microscopy. Immunity. 2004;21:349–355. doi: 10.1016/j.immuni.2004.08.005. [DOI] [PubMed] [Google Scholar]
  • 127.Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427:355–360. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
  • 128.Schwab SR, Pereira JP, Matloubian M, et al. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science. 2005;309:1735–1739. doi: 10.1126/science.1113640. [DOI] [PubMed] [Google Scholar]
  • 129.Zachariah MA, Cyster JG. Neural crest-derived pericytes promote egress of mature thymocytes at the corticomedullary junction. Science. 2010;328:1129–1135. doi: 10.1126/science.1188222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Shiow LR, Rosen DB, Brdicková N, et al. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature. 2006;440:540–544. doi: 10.1038/nature04606. [DOI] [PubMed] [Google Scholar]
  • 131.Mandala S, Hajdu R, Bergstrom J, et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296:346–349. doi: 10.1126/science.1070238. [DOI] [PubMed] [Google Scholar]
  • 132.Brinkmann V. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J Biol Chem. 2002;277:21453–21457. doi: 10.1074/jbc.C200176200. [DOI] [PubMed] [Google Scholar]
  • 133.Brinkmann V. FTY720 (fingolimod) in multiple sclerosis: therapeutic effects in the immune and the central nervous system. Br J Pharmacol. 2009;158:1173–1182. doi: 10.1111/j.1476-5381.2009.00451.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chi H. Sphingosine-1-phosphate and immune regulation: trafficking and beyond. Trends Pharmacol Sci. 2011;32:16–24. doi: 10.1016/j.tips.2010.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mehling MM, Brinkmann VV, Antel JJ, et al. FTY720 therapy exerts differential effects on T cell subsets in multiple sclerosis. Neurology. 2008;71:1261–1267. doi: 10.1212/01.wnl.0000327609.57688.ea. [DOI] [PubMed] [Google Scholar]
  • 136.Pappu R, Schwab SR, Cornelissen I, et al. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science. 2007;316:295–298. doi: 10.1126/science.1139221. [DOI] [PubMed] [Google Scholar]
  • 137.Drumea-Mirancea M. Characterization of a conduit system containing laminin-5 in the human thymus: a potential transport system for small molecules. J Cell Sci. 2006;119:1396–1405. doi: 10.1242/jcs.02840. [DOI] [PubMed] [Google Scholar]
  • 138.Bearman RMR, Bensch KGK, Levine GDG. The normal human thymic vasculature: an ultrastructural study. Anat Rec. 1975;183:485–497. doi: 10.1002/ar.1091830402. [DOI] [PubMed] [Google Scholar]
  • 139.Vianello F, Kraft P, Mok YT, et al. A CXCR4-dependent chemorepellent signal contributes to the emigration of mature single-positive CD4 cells from the fetal thymus. J Immunol. 2005;175:5115–5125. doi: 10.4049/jimmunol.175.8.5115. [DOI] [PubMed] [Google Scholar]
  • 140.Poznansky MC, Olszak IT, Foxall R, et al. Active movement of T cells away from a chemokine. Nat Med. 2000;6:543–548. doi: 10.1038/75022. [DOI] [PubMed] [Google Scholar]
  • 141.Shiow LR, Roadcap DW, Paris K, et al. The actin regulator coronin 1A is mutant in a thymic egress-deficient mouse strain and in a patient with severe combined immunodeficiency. Nat Immunol. 2008;9:1307–1315. doi: 10.1038/ni.1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Moshous D, Martin E, Carpentier W, et al. Whole-exome sequencing identifies coronin-1A deficiency in 3 siblings with immunodeficiency and EBV-associated B-cell lymphoproliferation. J Allergy Clin Immunol. 2013;131:1594e9–1603e9. doi: 10.1016/j.jaci.2013.01.042. [DOI] [PubMed] [Google Scholar]
  • 143.Mou F, Praskova M, Xia F, et al. The Mst1 and Mst2 kinases control activation of rho family GTPases and thymic egress of mature thymocytes. J Exp Med. 2012;209:741–759. doi: 10.1084/jem.20111692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Milner JD, Holland SM. The cup runneth over: lessons from the ever-expanding pool of primary immunodeficiency diseases. Nat Rev Immunol. 2013;13:635–648. doi: 10.1038/nri3493. [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES